PICK1 Deficiency Exacerbates Sepsis-Associated Acu

You might also like

You are on page 1of 12

Hindawi

BioMed Research International


Volume 2021, Article ID 9884297, 12 pages
https://doi.org/10.1155/2021/9884297

Research Article
PICK1 Deficiency Exacerbates Sepsis-Associated Acute
Kidney Injury

Qian Dou ,1 Hang Tong ,2 Yichun Yang,3 Han Zhang,1 and Hua Gan 1

1
Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
2
Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
3
Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China

Correspondence should be addressed to Hua Gan; ganhua113@sohu.com

Qian Dou and Hang Tong contributed equally to this work.

Received 27 April 2021; Revised 19 June 2021; Accepted 24 June 2021; Published 9 July 2021

Academic Editor: Dorota Formanowicz

Copyright © 2021 Qian Dou et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

We performed in vitro and in vivo experiments to explore the role of protein kinase C-binding protein 1 (PICK1), an intracellular
transporter involved in oxidative stress-related neuronal diseases, in sepsis-related acute kidney injury (AKI). Firstly, PCR, western
blotting, and immunohistochemistry were used to observe the expression of PICK1 after lipopolysaccharide- (LPS-) induced AKI.
Secondly, by inhibiting PICK1 in vivo and silencing PICK1 in vitro, we further explored the effect of PICK1 on AKI. Finally, the
relationship between PICK1 and oxidative stress and the related mechanisms were explored. We found that the expression of
PICK1 was increased in LPS-induced AKI models both in vitro and in vivo. PICK1 silencing significantly aggravated LPS-
induced apoptosis, accompanied by ROS production in renal tubular epithelial cells. FSC231, a PICK1-specific inhibitor,
aggravated LPS-induced kidney injury. Besides, NAC (N-acetylcysteine), a potent ROS scavenger, significantly inhibited the
PICK1-silencing-induced apoptosis. In conclusion, PICK1 might protect renal tubular epithelial cells from LPS-induced
apoptosis by reducing excessive ROS, making PICK1 a promising preventive target in LPS-induced AKI.

1. Introduction to understand the potential mechanism of LPS-induced renal


tubular cell injury and apoptosis.
Acute kidney injury (AKI) is common in hospitalized patients, Reactive oxygen species (ROS), which contained exoge-
with various complications and high mortality, and brings a nous oxidants and active oxygenated compounds produced
great challenge for clinicians [1–3]. Sepsis is the most common in oxidative metabolism in vivo, plays an essential role in cell
cause of AKI in critically ill patients, [4] and lipopolysaccha- apoptosis [10, 11]. When cells are exposed to harmful stimu-
ride (LPS) is an important initiating factor of sepsis, which lation, a lot of ROS will be produced, leading to the oxidant-
can induce cytokine storm, oxidative stress, hypotension, antioxidant imbalance [12–14]. Recent studies have shown
insufficient renal perfusion, premature senescence, and even- that oxidative stress is the leading cause of LPS-induced
tually lead to the gradual decline of renal function, [5, 6] thus mitochondrial damage in renal cells [15], and the mitochon-
participating in the occurrence and development of sepsis- drial dysfunction in turn triggers apoptosis by activating the
associated acute kidney injury (SA-AKI). LPS would make a cascade of tandem proteases [16–19]. Therefore, reducing
valuable contribution towards the pathogenesis of sepsis- oxidative stress may be an effective way to decrease LPS-
associated AKI. Moreover, apoptosis of renal tubular epithelial induced apoptosis of TECs.
cells (TECs) also contributes to the progress of AKI [7–9]. Protein interacting with C-kinase 1 (PICK1) is a unique
Apoptosis induced by endotoxin plays an important role in protein containing both BAR (Bin/Amphiphysin/Rvs) and
sepsis-associated acute renal injury. Therefore, it is necessary PDZ (PSD-95/DlgA/ZO-1) domains. This particular struc-
2 BioMed Research International

ture enables PICK1 to bind to various membrane proteins, we complete the remaining dyeing steps according to the
thus initiating multiple functions [20–22]. PICK1 is widely manufacturer’s immunoassay kit instructions (ZSGB-BIO
expressed in various organs and has been found to participate Technology Co., Ltd., Beijing, China). PICK1 (10983-2-AP,
in the oxidative regulation by regulating glutathione (GSH, Proteintech, China) and cleaved caspase 3 (19677-1-AP,
an important antioxidant) [23] in the lung [24] and liver Proteintech, China) were used as the primary antibody at a
[25]. However, the research on the relationship between dilution of 1 : 200. The images were taken with a digital
PICK1 and SA-AKI is relatively rare. camera under a microscope.
In our study, using a septic mouse model by injecting
LPS, we investigated the role of PICK1 in the pathophysio- 2.5. Cell Culture and Treatment. The human renal proximal
logical process of septic-induced AKI and explored the tubular epithelial cell line (HK2) was presented by Dr. Jiang,
underlying mechanism. Center for kidney disease, Chongqing Medical University.
The cells were cultured in 1640 medium (Gibco, USA)
2. Materials and Methods containing 10% FBS (PAN-Biotech, Adenbach, Germany)
and 1% penicillin-streptomycin (Beyotime, China) at 37°C
2.1. Animal Models and Groups. Our animal experiment was in 5% CO2 wet air with or without Lenti-NC and Lenti-
carried out in the Animal Laboratory Center of Chongqing PICK1 to silence the expression of PICK1 (NC and sh-
Medical University (Chongqing, China). Male C57BL/6 mice PICK1). And then, for the treatment with LPS (15 μg/ml)
(6–8 weeks old, 20–25 g) purchased from the Animal Labora- for 24 hours, the cells were divided into the WT group,
tory Center of The First Affiliated Hospital of Chongqing WT+LPS group, NC+LPS group, and sh-PICK1+LPS group
Medical University were used in this study. The Animal for further experiments.
Experimentation Ethics Committee of The First Affiliated
Hospital of Chongqing Medical University approved all 2.6. Transfection. Lenti-NC and Lenti-PICK1 were built at
animal experiments. Mice were housed under specific Shanghai Genechem Co., Ltd. (Shanghai, China). HK2 cells
pathogen-free conditions, and the standard rodent chow were seeded into 6-well plates. When the cell fusion reached
and water were available ad libitum. The septic AKI animal 50%, the medium containing lentivirus and HitransG A was
model was established by intraperitoneal injection of 10 added. After 24 hours of incubation, the medium was
mg/kg of LPS for 24 hours [15, 26]. changed, and the cells were cultured for another 24 hours
FSC231 (529531, Merck, Germany), a specific PICK1 for further analysis. Besides, stably transfected cell lines were
inhibitor [27], was given to examine the role of PICK1 in screened by puromycin (2 ng/ml). Then, cells were cultured
LPS-induced AKI. C57BL/6 mice were randomly divided with the optimum concentration of LPS for the given time.
into three groups (n = 6 in each group): the control group,
LPS group, and LPS+FSC231 group. The LPS group was 2.7. Immunofluorescence Staining. HK2 were inoculated in 24
given intraperitoneal injection of LPS, and the control group aperture plates after the cells reached 40% to 50% confluency,
was given the same dose of saline. The LPS+FSC231 group treated with LPS for 24 hours after adhering the wall, fixed
was injected with FSC231 (78.4 μg/g) for five consecutive with 4% paraformaldehyde for 15 minutes at room tempera-
days, followed by LPS administration 2 hours after the last ture, and then immersed with 0.1% TritonX-100 (p0096,
dose of FSC231 [25]. Renal tissue and blood samples were Jiangsu, China) and 5% goat serum for 15 min and 30 min,
collected after 24 hours of LPS treatment. respectively. After culture with PICK1 antibody (1 : 200, Pro-
teintech, China) at 4°C overnight, goat anti-rabbit IgG (1 : 20,
2.2. Assessment of Kidney Function. Blood samples were col-
Proteintech, China) was added in the dark for 1 hour. The
lected from the eyeballs. According to the manufacturer’s
cells were then stained with 2-(4-aminophenyl)-6-indolea-
illustration, blood urea nitrogen (BUN) and serum creatinine
mine dihydrochloride (DAPI) for 10 minutes. Zeiss LSM
(Scr) were measured enzymatically.
800 confocal laser scanning microscope (Carl Zeiss,
2.3. Histology Analyses. Kidney specimens were fixed with 4% Germany) was used for image acquisition and observation.
paraformaldehyde, embedded in paraffin, sectioned into 4
μm thick, and stained with hematoxylin and eosin. The path- 2.8. GSH Measurement. According to the manufacturer’s
ological changes of renal tissue were observed under the light illustration, GSH-Px (Total Glutathione Peroxidase Test
microscope. A semiquantitative scoring method was used to Kit, S0053, Beyotime, China) was determined with the corre-
evaluate the degree of renal tubular injury: 0 for less than sponding test kit.
5%, 1 for 5%-25%, 2 for 25%-50%, 3 for 50%-75%, and 4
for more than 75%. Ten areas in each section were randomly 2.9. Detection of Intracellular ROS Levels. According to the
observed under the light microscope (400x magnification) to manufacturer’s illustration, ROS was determined with the
evaluate tubular epithelial necrosis, lumen dilatation, and corresponding test kit (Reactive Oxygen Species Assay Kit,
tubular injury. S0053, Beyotime, China). In short, the treated cells were
incubated with DCFH-DA (10 μmol/l) in the dark at 37°C
2.4. Immunohistochemistry. The expression of PICK1 and for 30 minutes and then washed with serum-free medium.
cleaved caspase 3 were detected by immunohistochemistry. The fluorescence was observed under flow cytometry. The
The dewaxed slices were put into citrate buffer (0.01 M, pH excitation and emission wavelengths were 488 nm and
6.0) and heated in microwave oven for 20 minutes. Then, 525 nm, respectively.
BioMed Research International 3

2.10. TUNEL Staining. According to the manufacturer’s 2.15. CCK-8. According to the manufacturer’s illustration,
instructions, the apoptotic cells in renal tissue were evaluated the proliferation of pretreatment cell samples was detected
with the TACS TdT In Situ Apoptosis Detection kit (Roche, by the CCK-8 kit (C0038, Beyotime, China). We add 10 μl
Germany). Renal tissue sections were deparaffinized and reagent to each sample for two hours and then detect the
hydrated as per the standard procedure and digested with absorbance at 450 nm wavelength.
proteinase K solution at 37°C for 30 min. After the endoge-
nous peroxidase activity was quenched, the sections were 2.16. Statistical Analysis. All data were expressed as the
incubated with TdT labeling reaction mixture at 37°C for means ± standard deviation and analyzed by GraphPad
one hour and then treated with streptavidin horseradish per- Prism 9.0 software using one-way ANOVA or t-test. Differ-
oxidase. Subsequently, the prepared samples were washed ences were considered significant at P < 0:05.
with phosphate buffer for colorimetric analysis. An optical
microscope was used to analyze the results. 3. Results
3.1. PICK1 Was Increased in the LPS-Induced Renal Proximal
2.11. Hoechst 33258 Staining. According to the manufac- Tubular Cells. Western blotting and qPCR were used to inves-
turer’s instructions, the cells were washed twice with PBS. tigate the effect of LPS on the expression of PICK1 in vitro. We
Then, the samples were fixed with 4% formaldehyde for found that the expression of PICK1 increased after LPS treat-
15 min at room temperature and stained with Hoechst ment at different concentrations (0, 5, 15, and 20 μg/ml,
33258 (Beyotime, China) solution in the dark for 5 minutes. Figures 1(a) and 1(b)). In HK2 cells, PICK1 gradually increased
After incubation, the apoptotic cells were observed under a with time after LPS treatment (15 μg/ml, Figure 1(c)). During
fluorescence microscope. our observation period, the expression of PICK1 reached the
peak after treatment with 15 μg/ml LPS for 24 hours. Therefore,
2.12. Real-Time Quantitative Polymerase Chain Reaction LPS induced the increasing expression of PICK1 protein in a
(RT-PCR). According to the manufacturer’s instructions, time- and dose-dependent manner.
cell RNA was extracted with TRIzol reagent (Invitrogen, The level of PICK1 in LPS-treated mouse kidney was also
Shanghai, China). Total RNA was reverse transcribed into detected in vivo. After treatment with LPS (10 mg/kg) for 24
cDNA by Prime Script RT Kit (Takara Biotechnology Inc., h, the expression of PICK1 in the renal cortex and medulla
Shiga, Japan). RT-qPCR was performed with the following significantly increased as showed in the immunohistochemi-
primer pairs: cal staining image (Figures 1(d) and 1(e)). The bands of
PICK1-F 5 ′ -AGTTCGGCATTCGGCTTC-3 ′ , PICK1-R western blotting also showed that PICK1 in the kidney tissue
5 ′ -GAAGCCGAATGCCGAACT-3 ′ , GAPDH-F 5 ′ -GGTG of LPS-induced AKI mice was increased, and FSC231
AAGGTCGGAGTCAACG-3 ′ , and GAPDH-R 5 ′ -CAAA decreased the level of PICK1 (Figure 1(f)).
GTTGTCATGGATGHACC-3 ′ .
3.2. Inhibition of PICK1 Aggravated the LPS-Induced AKI.
The target gene level was normalized with GAPDH level,
FSC231, acting as a particular PICK1 inhibitor [27], was
and the mRNA level was calculated by the standard method.
administered to investigate the role of PICK1 in LPS-
induced AKI. The immunohistochemistry results showed
2.13. Western Blot. RIPA buffer containing PMSF and phos- that FSC231 reduced the PICK1 expression in LPS-induced
phatase inhibitors was used to extract the proteins of renal AKI (Figures 2(a) and 2(b)). Both BUN and serum creatinine
tissue and HK2 cells protein (all from Beyotime, China). (Scr) presented a rapid increase after FSC231 treatment
According to the manufacturer’s protocol, the protein (Figures 2(c) and 2(d)). Histological analysis revealed that
concentration was determined by the BCA protein assay the expression of PICK1 in the kidney tissue of LPS-
kit. The protein samples of each group were separated by induced AKI mice was increased, and FSC231 treatment
SDS-PAGE and transferred to PVDF membranes (MILI aggravated AKI, which was reflected by the exfoliated TECs,
Bloomberg, Massachusetts). The PVDF membranes were the damaged tubular structure, and the necrotic epithelial
sealed with rapid blocking solution at room temperature. cells (Figures 2(e) and 2(h)). The number of TUNEL-
The primary antibodies included rabbit anti PICK1 (1 : 500, positive cells in the kidney of mice pretreated with PICK1
protein tech,) rabbit anti Cleaved Caspase-3, rabbit anti inhibitor FSC231 was also increased (Figures 2(f) and 2(g)).
Bax, and rabbit anti BCL-2 (1 : 500, Proteintech, China), rab- Therefore, the inhibition of PICK1 aggravated the LPS-
bit anti ASK1 (1 : 500,Proteintech, China), rabbit anti induced AKI in vivo. However, the appeal results all show
p38MAPK (1 : 500, Proteintech, China), and rabbit anti that using FSC231 alone without LPS have no significant
GAPDH (1 : 8000; Proteintech, China). Using the HRP- effect on tissue morphology and function.
labeled secondary antibody (1 : 3000 dilution) to incubate
the membrane at 37°C for 1 hour. Finally, the fusion imaging 3.3. PICK1 Silencing Promoted TEC Apoptosis. Compared
system is used to detect the relative densities of the bands. with the control group, after 24 hours of transfecting, PICK1
shRNA significantly reduced both the mRNA and protein of
2.14. Flow Cytometry. Annexin V-FITC/PC was used to PICK1 (Figures 3(a) and 3(b)). The proapoptotic protein Bax
detect apoptosis with the test kit (Beyotime, China) accord- and cleaved-caspase 3 was upregulated while the antiapopto-
ing to the manufacturer’s instructions. The apoptosis was tic protein Bcl-2 was downregulated after PICK1 silencing
analyzed by flow cytometry (BD Biosciences, USA). (Figure 3(c)). Similarly, the expression of cleaved-caspase 3
4 BioMed Research International

LPS (μg/ml) 0 5 15 20
PICK1 50kDa
2.5 ns Time (h) 0 12 24 48
⁎⁎⁎ GAPDH 36kDa PICK1 50kDa
2.0 2.0 GAPDH 36kDa
Relative PICK1 level

ns 2.0 ns

Protein related level


(Protein/GAPDH)
⁎⁎⁎

Protein-related level
1.5 ⁎⁎⁎

(Protein/GAPDH)
1.5
1.5
1.0 1.0
1.0
ns ⁎
0.5 0.5 0.5

0.0 0.0 0.0


LPS (μg/ml) 0 5 15 20 PICK1 PICK1

LPS (μg/ml) 0h 24h


0 15 12h 48h
5 20
(a) (b) (c)
LPS - + +
FSC231 - - +

60 PICK1-Vivo PICK1
⁎⁎ Vivo
GAPDH
⁎⁎⁎
PICK1 IHC score

40 1.0 ⁎⁎

Protein-related level
(Protein/GAPDH)
0.8
PICK1-Vivo
0.6
20
0.4
0.2
20um 20um
0 0.0
Control LPS Control LPS PICK1
Control
LPS
LPS+FSC231
(d) (e) (f)

Figure 1: PICK1 was increased in LPS-induced AKI models both in vitro and in vivo. (a, b) PCR and western blotting results after treatment
with different concentrations of LPS for 24 hours (0 μg/ml versus 5 μg/ml, 0 μg/ml versus 15 μg/ml; ∗ P < 0:05, ∗∗∗ P < 0:001). (c) Western
blotting results after LPS treatment for different times (0 h versus 12 h, 0 h versus 24 h; ∗ P < 0:05, ∗∗∗ P < 0:001). (d, e) Representative
immunohistochemical images of pretreated renal tissue (magnification ×400, scale = 20 μm, the control versus LPS, ∗∗ P < 0:01). (f)
Representative western blotting analysis showed the expression of PICK1 in the kidney of mice treated with LPS (10 mg/kg) and FSC231
(78.4 μg/g) (the control versus LPS, ∗∗ P < 0:01).

in the LPS+FSC231 group was significantly increased in trations (Figure 3(i)). These results indicated that PICK1 was
immunohistochemistry, but in the case of using FSC231 involved in the proapoptotic effect of LPS on HK2 cells.
alone, the expression of cleaved-caspase 3 did not change sig-
nificantly (Figure 3(d)), suggesting that under the conditions 3.4. PICK1 Inhibition Increased the Production of Peroxide
of LPS-induced septic kidney injury, PICK1 inhibition aggra- and Activated the ASK1-p38 Apoptotic Pathway after LPS
vated the apoptosis of renal cells. The immunofluorescence Treatment. As an important intracellular regulatory metabo-
results of HK2 cells showed that LPS could increase the lite, GSH could act as a direct antioxidant to participate in
expression of PICK1, but shRNA could decrease PICK1 biotransformation. We detected the changes in GSH and
(Figures 3(e) and 3(g)). Hoechst 33258 staining showed that ROS content after LPS pretreatment. GSH measurement
PICK1 silencing evidently exacerbated LPS-induced apopto- result showed that the content of GSH in the PICK1 inhibi-
sis (Figures 3(f) and 3(h)). Moreover, the CCK-8 test showed tion group was significantly lower than that in the NC group
that the proliferation of cells in the PICK1 knockdown group both in vivo and in vitro (Figures 4(a) and 4(b)). The results
was significantly slowed down at different times and concen- of flow cytometry indicated that the peroxide product in the
BioMed Research International 5

60 PICK1-Vivo

PICK1 IHC scores


PICK1-Vivo
40 ⁎

20

20um 20um
0
LPS LPS+FSC231
LPS LPS+FSC231
(a) (b)
60 BUN 30 Scr
⁎⁎ ⁎⁎

⁎ ⁎⁎
40 20
BUN (mmol/l)

Scr (μmol/l)
ns ns

20 10

0 0
Control FSC231 LPS LPS+231 Control FSC231 LPS LPS+231

(c) (d)
LPS - - + +
FSC231 - + - +

HE

20um 20um 20um 20um

(e)
LPS - - + +
FSC231 - + - +

TUNEL

20um 20um 20um 20um

(f)
250 5
⁎⁎ ⁎⁎⁎
TUNEL-positive nuclei

200 4
Tubular injury scores

⁎⁎⁎ ⁎⁎⁎
150 3

100 2
ns ns
50 1

0 0
Control FSC231 LPS LPS+Fsc231 Control FSC231 LPS LPS+FSC231

(g) (h)

Figure 2: PICK1 inhibition aggravated LPS-induced AKI. (a, b) The results of immunohistochemistry (magnification ×400, scale = 20 μm,
LPS versus LPS+FSC231, ∗ P < 0:05). (c) The results of BUN (the control versus LPS, LPS versus LPS+FSC231; ∗∗∗ P < 0:001). (d) The
results of Scr (the control versus LPS, LPS versus LPS+FSC231, ∗∗ P < 0:01). (e) Hematoxylin-eosin (H & E) staining showed the renal
histological damage (magnification ×400, scale = 20 μm). (f, g) Representative images and statistical analysis of TUNEL-positive cells
(magnification ×400, scale = 20 μm, the control versus LPS, LPS versus LPS+FSC231; ∗∗ P < 0:01, ∗∗∗ P < 0:001). (h) The results of tubular
injury scores (the control versus LPS, LPS versus LPS+FSC231, ∗∗∗ P < 0:001).
6 BioMed Research International

1.5

Relative PICK1 level


1.0 1.5
⁎⁎

Protein-related level
(Protein/GAPDH)
1.0
0.5
WT NC sh-PICK1 0.5
PICK1 50kDa

GAPDH 36kDa 0.0


0.0
PICK1
WT NC sh-PICK1
WT
NC
sh-PICK1

(a) (b)
LPS - - + +
FSC231 - + - +
WT NC sh-PICK1
Bax 21kDa

Cleaved- 19kDa
caspase 3 17kDa 20um 20um 20um 20um

Bcl-2 26kDa Cleaved-caspase 3


80 ⁎⁎
GAPDH 36kDa
Cleaved-caspase 3 IHC scores

2.5
⁎⁎⁎ 60
⁎⁎⁎
Protein-related level

2.0
(Protein/GAPDH)

⁎⁎⁎ ⁎⁎⁎
1.5 40
ns
ns
1.0
20
ns ns
0.5

0.0 0
Bax Cleaved- Bcl-2 Control FSC231 LPS LPS+FSC231
caspase 3

WT
NC
sh-PICK1

(c) (d)

WT WT+LPS NC+LPS sh-PICK1+LPS


(e)

Figure 3: Continued.
BioMed Research International 7

10 ⁎⁎⁎
ns

PICK1 relative red fluorescence


8

0
WT WT+LPS NC+LPS sh-PICK1+LPS

PS
LP

LP
W

+L
T+

C+

K1
W

IC
-P
Sh
(f) (g)
80
⁎⁎⁎

1.0
60
Cell viability (OD 450nm)
Apoptosis rate (%)

Cell viability (% of control)


0.8 120
40 100
0.6
ns 80
0.4 60
20
40
0.2
20
0 0.0 0
12 24 48 72 0 5 10 15
T

PS
LP

LP
W

+L
T+

C+

K1

Time (hours) LPS Concentration (μg/ml)


W

IC
-P
Sh

Control Control
NC NC
sh-PICK1 sh-PICK1
(h) (i)

Figure 3: Downregulation of PICK1 aggravated LPS-induced apoptosis of HK2 cells. (a, b) The levels of PICK1 mRNA and protein were
detected by qPCR and WB (NC versus sh-PICK1, ∗∗ P < 0:01). (c) The expression levels of apoptosis-related proteins Bcl-2, Bax, and
cleaved caspase 3 were detected by western blotting (NC versus sh-PICK1, ∗∗∗ P < 0:001). (d) Representative cleaved caspase 3 level
detected by immunohistochemistry (the control versus LPS, LPS versus LPS+FSC231; ∗∗ P < 0:01, ∗∗∗ P < 0:001). (e, g) Representative
immunofluorescence results (WT+LPS versus NC+LPS, NC+LPS versus sh-PICK1+LPS; ∗∗∗ P < 0:001). (f–h) The apoptosis of HK2 cells
was detected by Hoechst 33258 staining (magnification ×200, scale = 50 μm, WT+LPS versus NC+LPS, NC+LPS versus sh-PICK1
+LPS; ∗∗∗ P < 0:001). (f, h) Representative Hoechst 33258 staining results (WT+LPS versus NC+LPS, NC+LPS versus sh-PICK1+LPS;
∗∗∗
P < 0:001). (i) The apoptosis of HK2 cells was detected by CCK-8.

sh-PICK1 group was significantly higher than that in the NC (Figures 5(a) and 5(b)). Additional pretreatment with 10
group (Figure 4(c)). Previous studies have shown that ROS mM NAC for one hour significantly downregulated the
can initiate apoptosis by activating the ASK1-p38MAPK PICK1-silencing-exacerbated ROS and TEC apoptosis
signaling pathway [44]. Our western blotting showed that (Figure 5(c)–5(e)). At the same time, the expression of
PICK1 inhibition upregulated the expression of ASK1 and ASK1 and p38MAPK was decreased, and the proapoptotic
p38MAPK both in the HK2 and mouse model, suggesting protein Bax and cleaved-caspase 3 was downregulated while
that PICK1 inhibition could activate the ASK1-p38MAPK the antiapoptotic protein Bcl-2 was upregulated after NAC
signaling pathway (Figures 4(d) and 4(e)). Therefore, PICK1 pretreatment detected by western blotting (Figures 5(f) and
silencing might be involved in LPS-induced ROS production 5(g)). Based on our results, ROS-mediated apoptosis may
and the activation of the ASK1-p38MAPK signaling path- participate in PICK1-inhibition-induced HK2 cell apoptosis
way in vitro. through activating the ASK1-p38MAPK signaling pathway.

3.5. NAC Inhibited the LPS-Induced Apoptosis and ROS. 4. Discussion


NAC is an important scavenger for oxidation products
in vivo [15]. We further explored the role of NAC on LPS- Acute kidney injury (AKI) is a significant challenge for clini-
induced apoptosis in TECs. Flow cytometry showed that cians, and sepsis-associated acute kidney injury (SA-AKI)
PICK1 silencing exacerbated LPS-induced early apoptosis has been proved to increase the mortality of children and
8 BioMed Research International

150 Vitro
60 Vivo

GSH concentration
ns

GSH concentration
100 ⁎⁎⁎
40 ⁎

(μm)

(μM)
50 20
⁎⁎⁎

0 0
NC sh-PICK1

31

31
ro

LP
C2

C2
nt
Co

FS

FS
l+

S+
ro

LP
nt
Co
(a) (b)
Vitro
WT NC sh-PICK1
ASK1 155kDa

p38MAPK 38kDa

GAPDH 36kDa
2.0 ⁎⁎⁎
400 400
Protein-related level ⁎⁎⁎
(Protein/GAPDH)
1.5
Count

Count

200 200 1.0

0.5 ns ns

0 0 0.0
102 103 104 105 106 107 102 103 104 105 106 107 ASK1 p38MAPK
DCFH FITC-A DCFH FITC-A
WT
NC sh-PICK1 NC
sh-PICK1
(c) (d)
2.0
⁎⁎⁎
Protein-related level
(Protein/GAPDH)

Vivo 1.5 ⁎⁎
LPS - - + +
FSC231 - + - + ⁎⁎⁎ ⁎⁎⁎
1.0
ASK1 155kDa
0.5
ns ns
p38MAPK 38kDa
0.0
ASK1 p38MAPK
GAPDH 36kDa
Control LPS
FSC231 LPS+FSC231
(e)

Figure 4: Knockdown of PICK1 increased the production of peroxide and activated the apoptotic pathway after LPS treatment. (a) After
treatment with LPS for 24 hours, the content of GSH in the NC and sh-PICK1 groups (NC versus sh-PICK1, ∗∗∗ P < 0:001). (b) The
content of GSH in each group of mouse model (LPS versus LPS+FSC231, ∗∗∗ P < 0:001). (c) Representative fluorescence images of
ROS. (d, e) Representative western images of the ASK1 and p38MAPK pathway (NC versus sh-PICK1, ∗∗ P < 0:01; ASK1: the
control versus LPS, ∗∗∗ P < 0:001; LPS versus LPS+FSC231, ∗∗ P < 0:01; p38MAPK: the control versus LPS, ∗∗∗ P < 0:001; LPS versus
LPS+FSC231, ∗∗∗ P < 0:001).

adults [1–3]. Endotoxin, especially lipopolysaccharide (LPS), nism of LPS-induced AKI, there is no effective intervention
is a common cause of SA-AKI [5]. However, since the lack of so far. PICK1 has been reported to have an antioxidant effect
understanding of the complex pathophysiological mecha- in the nervous system [23], but the role of PICK1 in AKI has
BioMed Research International 9

Q1-UL (1.21%) Q1-UR (6.60%) Q1-UL (0.73%) Q1-UR (9.43%) Q1-UL (0.61%) Q1-UR (12.91%) Q1-UL (2.84%) Q1-UR (23.10%)
106 106 106 106
DAPI PB450-A

DAPI PB450-A

DAPI PB450-A

DAPI PB450-A
105 105 105 105

104 104 104 104

103 103 103 103


Q1-LL (89.08%) Q1-LR (3.11%) Q1-LL (78.82%) Q1-LR (11.02%) Q1-LL (75.61%) Q1-LR (10.87%) Q1-LL (67.22%) Q1-LR (6.84%)

103 104 105 106 103 104 105 106 103 104 105 106 103 104 105 106
Annexin V APC-A Annexin V APC-A Annexin V APC-A Annexin V APC-A
WT WT+LPS NC+LPS sh-PICK1+LPS
(a)

0.4 400
Apoptosis 300
⁎⁎
Apoptotic rate (%)

0.3
200
⁎⁎
Count

Count
0.2 200

100
0.1

0.0 0 0
102 103 104 105 106 107 102 103 104 105 106 107
T

PS
LP

LP
W

+L

DCFH FITC-A DCFH FITC-A


T+

C+

K1
W

IC

sh-PICK1+LPS sh-PICK1+LPS+NAC
-P
sh

(b) (c)
0.3

Q1-UL (2.58%) Q1-UR (20.12%) Q1-UL (1.23%) Q1-UR (11.67%)


106 106
Apoptotic rate (%)

0.2
DAPI PB450-A

DAPI PB450-A

105 105 ⁎⁎⁎

104 104
0.1

103 103
Q1-LL (70.45%) Q1-LR (6.85%) Q1-LL (84.21%) Q1-LR (2.89%)
0.0
103 104 105 106 103 104 105 106
PS

AC

Annexin V APC-A Annexin V APC-A


+L

+N
K1

PS
IC

sh-PICK1+LPS sh-PICK1+LPS+NAC
+L
-P

K1
sh

IC
-P
sh

(d) (e)
1.0
Protein-related level
(Protein/GAPDH)

Vitro 0.8
sh-PICK1 + +
LPS + + 0.6 ⁎⁎
NAC - + 0.4 ⁎⁎⁎
ASk1 155kDa 0.2

p38MAPK 38kDa 0.0


ASK1 p38MAPK
GAPDH 36kDa
sh-PICK1+LPS
sh-PICK1+LPS+NAC
(f)

Figure 5: Continued.
10 BioMed Research International

1.5
Vitro

Protein-related level
(Protein/GAPDH)
sh-PICK1 + + ⁎⁎⁎
1.0
LPS + +
NAC - +
Bax 21kDa ⁎⁎⁎
0.5
⁎⁎⁎
Cleaved- 19kDa
caspase 3
17kDa 0.0
Bcl-2 26kDa Bax Cleaved- Bcl-2
caspase 3
GAPDH 36kDa
sh-PICK1+LPS
sh-PICK1+LPS+NAC

(g)

Figure 5: Inhibition of PICK1 expression could increase LPS-induced apoptosis, and NAC could alleviate pretreatment-induced apoptosis.
(a, b) After 24 hours of LPS treatment in the different groups, apoptosis rate was detected by flow cytometry (WT versus WT+LPS, NC+LPS
versus sh-PICK1+LPS; ∗∗ P < 0:01). (c–e) HK2 cells were pretreated with 10 mM NAC for 1 h and then treated with LPS for another 24 h.
Representative images of ROS and apoptosis rate in flow cytometry after NAC and LPS treatment (sh-PICK1+LPS versus sh-PICK1+LPS
+NAC, ∗∗∗ P < 0:001). (f) After treatment with NAC, the ASK1 and p38MAPK were detected by western blotting (sh-PICK1+LPS versus
sh-PICK1+LPS+NAC; ∗∗ P < 0:01, ∗∗∗ P < 0:001). (f) After treatment with NAC, the apoptotic protein was detected by western blotting
(sh-PICK1+LPS versus sh-PICK1+LPS+NAC, ∗∗∗ P < 0:001).

barely been reported. Thus, we investigated whether PICK1 chondrial dysfunction, which caused a large-scale production
played a vital role in LPS-induced AKI in vivo and in vitro of reactive oxygen species (ROS). JNK and p38MAPK,
and found that PICK1 deficiency exacerbated SA-AKI. members of the MAPKs family, could transmit apoptosis
Apoptosis is a programmed cell death necessary for cellu- signal to the mitochondria, release cytochrome C, activate
lar homeostasis [28]; however, excessive apoptosis can also specific caspase enzyme, and induce apoptosis.
cause tissue damage [29, 30]. The regulation of apoptosis Previous studies have shown that PICK1 interacts with
involves many steps, and there are 3 main apoptosis regulat- various neurotransmitter receptors, enzymes, and trans-
ing pathways as follows: the exogenous pathway, in which the porters through its unique structure to affect synaptic
death receptor pathway is a very important part [31]; in the function, leading to nerve damage, such as epilepsy and Par-
intrinsic pathway, in which the mitochondrial apoptotic kinson’s disease [22, 23, 40]. PICK1 was also involved in
signaling pathway plays a pivotal role [32]; and in the endo- breast cancer by inhibiting TGF-β signaling, thus initiating
plasmic reticulum stress-induced pathway, in which ROS early cancer [41]. Besides, PICK1 deficiency aggravated
destroyed the ER function and initiated unfolded protein sepsis-induced acute lung injury through lysosomal injury
response (UPR) and endoplasmic reticulum stress (ERS) [24, 42]. And PICK1 may mediate the prosurvival activity
in vivo and vitro, which might be an important mechanism of PKCalpha by serving as a molecular link between PKCal-
that lead to tissue damage and cell apoptosis [33]. This study pha and the mitochondria, which results in a more stable
found that PICK1 inhibitor FSC231 significantly promoted mitochondrial membrane potential, enhances phosphoryla-
the apoptosis of renal tubular epithelial cells in vivo and tion of the anti-apoptotic Bcl-2 protein, and decreases
aggravated the LPS-induced renal injury. However, the use dimerization of the.
of FSC231 alone has no obvious effect on the functions of proapoptotic Bax protein [45]. In other words, PICK1
the kidney tissue. Therefore, PICK1 has a protective effect was involved in the pathophysiological changes of many
on AKI induced by LPS. diseases. Recently, PICK1 has been reported to regulate glu-
Oxidative stress is an inevitable reaction in life [34]. tathione (GSH) homeostasis, indicating that PICK1 may play
Various harmful stimuli can break the balance of oxidative a key role in oxidative stress [23]. However, the relationship
stress, leading to cell apoptosis and pathological damage between PICK1 and LPS-induced AKI remains unclear.
[12–14]. Previous studies have shown that oxidative stress In our research, PICK1 acted as a negative regulator of
played a vital role in the pathogenesis of septic kidney injury ROS production in the mitochondria by regulating GSH
[15]. Oxidative stress and its induced apoptosis were con- content and influenced the intrinsic apoptotic pathway to
firmed to be essential pathogenic factors in chronic renal fail- improve cell survival. We used sh-PICK1 to inhibit PICK1
ure (CRF), [35] renal ischemia-reperfusion injury(IR) [36], expression in HK2 to further explore whether PICK1 could
acute renal injury (AKI) [37], and diabetic nephropathy protect TECs in septic conditions. Our results demonstrated
(DN) [38]. Among various mechanisms, the endogenous that PICK1 was upregulated after LPS treatment. Silencing
pathway of mitochondrial initiation; the regulation of the PICK1 increased cell and tissue damage. According to the
Bcl-2 family, NF-κB, and MAPK family; and the activation Hoechst staining, flow cytometry, and western blotting
of caspase were most closely related to oxidative stress- results, knockout of PICK1 raised LPS-induced ROS produc-
induced apoptosis [39]. In our septic AKI model, we found tion and aggravated the apoptosis of renal TECs. We further
that oxidative stress was the main factor leading to mito- confirmed that ROS production activated the ASK1-
BioMed Research International 11

p38MAPK apoptosis pathway and eventually led to cell and [3] R. Alobaidi, R. K. Basu, S. L. Goldstein, and S. M. Bagshaw,
tissue damage. Therefore, PICK1 may play an essential role “Sepsis-associated acute kidney injury,” Seminars in Nephrol-
in LPS-induced renal TEC apoptosis. Similarly, immunohis- ogy, vol. 35, no. 1, pp. 2–11, 2015.
tochemistry and pathological sections showed that FSC231 [4] C. L. Manrique-Caballero, G. Del Rio-Pertuz, and H. Gomez,
could increase the expression of an apoptotic protein in renal “Sepsis-associated acute kidney injury,” Critical Care Clinics,
cells and aggravate renal tissue injury. Both in vitro and vol. 37, no. 2, pp. 279–301, 2021.
in vivo experiments showed that silencing PICK1 reduced [5] D. H. Kim, Y. J. Jung, A. S. Lee et al., “COMP-angiopoie-
the production of GSH but increased the ROS in the LPS- tin-1 decreases lipopolysaccharide-induced acute kidney
induced AKI model. NAC is a kind of ROS scavenger [15, injury,” Kidney International, vol. 76, no. 11, pp. 1180–
43]. Pretreatment with NAC significantly decreased apopto- 1191, 2009.
sis and ROS and alleviated the PICK1-inhibition-induced [6] C. Chen, R. Qiu, J. Yang et al., “Lipoxin A4 restores septic renal
injury of HK2 cells. Based on these results, PICK1 silencing function via blocking crosstalk between inflammation and pre-
mature senescence,” Frontiers in Immunology, vol. 12,
upregulated ROS production by decreasing GSH content,
p. 637753, 2021.
[26] which might explain the mechanism of PICK1-
mediated cell survival. [7] A. Agarwal, Z. Dong, R. Harris et al., “Cellular and molecular
mechanisms of AKI,” Journal of the American Society of
In conclusion, we first demonstrated that the highly
Nephrology: JASN, vol. 27, no. 5, pp. 1288–1299, 2016.
expressed PICK1 after LPS-induced AKI might be an
[8] A. Koçkara and M. Kayataş, “Renal cell apoptosis and new
endogenous protective factor. Simultaneously, the ASK1-
treatment options in sepsis-induced acute kidney injury,”
p38MAPK pathway might be involved in the promotion Renal Failure, vol. 35, no. 2, pp. 291–294, 2013.
of PICK1-deficiency-induced apoptosis. Specifically, the
[9] J. Y. Kim, J. Leem, and K. K. Park, “Antioxidative, antiapopto-
silencing of PICK1 increased the production of ROS and
tic, and anti-inflammatory effects of apamin in a murine
activated the apoptotic pathway, thus aggravating the apo- model of lipopolysaccharide-induced acute kidney injury,”
ptosis of HK2 cells. Therefore, PICK1 may be a promising Molecules : a journal of synthetic chemistry and natural product
preventive target in LPS-induced AKI. chemistry, vol. 25, no. 23, p. 5717, 2020.
[10] M. Li, C. M. Li, Z. C. Ye et al., “Sirt3 modulates fatty acid oxi-
Data Availability dation and attenuates cisplatin-induced AKI in mice,” Journal
of Cellular and Molecular Medicine, vol. 24, no. 9, pp. 5109–
All supporting data are included within the main article. 5121, 2020.
[11] B. Zhang, Y. Wang, Y. Shan, and W. Yin, “Acetylharpagide
Conflicts of Interest protects mice from Staphylococcus Aureus-Induced acute
Lung injury by inhibiting NF-κB signaling pathway,” Mole-
The authors declare that the research was conducted in the cules, vol. 25, no. 23, p. 5523, 2020.
absence of any commercial or financial relationships that [12] U. Z. Paracha, K. Fatima, M. Alqahtani et al., “Oxidative stress
could be construed as a potential conflict of interest. and hepatitis C virus,” Virology Journal, vol. 10, no. 1, p. 251,
2013.
Authors’ Contributions [13] J. Choi and J. H. James Ou, “Mechanisms of liver injury. III.
Oxidative stress in the pathogenesis of hepatitis C virus,”
GH and DQ designed the research and revised the manuscript. American Journal of Physiology. Gastrointestinal and Liver
DQ and TH performed the experiments and wrote the draft Physiology, vol. 290, no. 5, pp. G847–G851, 2006.
manuscript. DQ, ZH, YYC and TH analyzed the experimental [14] P. Pavlakou, V. Liakopoulos, T. Eleftheriadis, M. Mitsis, and
results. All authors read and approved the manuscript and E. Dounousi, “Oxidative Stress and Acute Kidney Injury in
Critical Illness: Pathophysiologic Mechanisms—Biomarker-
agree to be accountable for all aspects of the research in ensur-
s—Interventions, and Future Perspectives,” Oxidative Medi-
ing that the accuracy or integrity of any part of the work are cine and Cellular Longevity, vol. 2017, Article ID 6193694, 11
appropriately investigated and resolved. Qian Dou and Hang pages, 2017.
Tong are co-first authors of the article.
[15] X. Zhong, J. He, X. Zhang et al., “UCP2 alleviates tubular
epithelial cell apoptosis in lipopolysaccharide- induced acute
Acknowledgments kidney injury by decreasing ROS production,” Biomedicine
& Pharmacotherapy, vol. 115, p. 108914, 2019.
We appreciate the Nephrology Department of The First [16] C. Brooks, Q. Wei, S. G. Cho, and Z. Dong, “Regulation of
Affiliated Hospital of Chongqing Medical University for the mitochondrial dynamics in acute kidney injury in cell culture
technical support. and rodent models,” The Journal of Clinical Investigation,
vol. 119, no. 5, pp. 1275–1285, 2009.
References [17] C. Quoilin, A. Mouithys-Mickalad, S. Lécart, M. P. Fontaine-
Aupart, and M. Hoebeke, “Evidence of oxidative stress and
[1] J. T. Poston and J. L. Koyner, “Sepsis associated acute kidney mitochondrial respiratory chain dysfunction in an in vitro
injury,” BMJ, vol. 364, p. k4891, 2019. model of sepsis-induced kidney injury,” Biochimica et Biophy-
[2] R. W. Schrier and W. Wang, “Acute renal failure and sepsis,” sica Acta, vol. 1837, no. 10, pp. 1790–1800, 2014.
The New England Journal of Medicine, vol. 351, no. 2, [18] E. Pathak, L. A. MacMillan-Crow, and P. R. Mayeux, “Role of
pp. 159–169, 2004. mitochondrial oxidants in an in vitro model of sepsis-induced
12 BioMed Research International

renal injury,” The Journal of Pharmacology and Experimental Complementary and Alternative Medicine, vol. 2020, Article
Therapeutics, vol. 340, no. 1, pp. 192–201, 2012. ID 8909171, 11 pages, 2020.
[19] S. M. Parikh, Y. Yang, L. He, C. Tang, M. Zhan, and Z. Dong, [35] G. Coppolino, G. Leonardi, M. Andreucci, and D. Bolignano,
“Mitochondrial function and disturbances in the septic kid- “Oxidative stress and kidney function: a brief update,” Current
ney,” Seminars in Nephrology, vol. 35, no. 1, pp. 108–119, 2015. Pharmaceutical Design, vol. 24, no. 40, pp. 4794–4799, 2018.
[20] J. Staudinger, J. Zhou, R. Burgess, S. J. Elledge, and E. N. Olson, [36] L. Gong, J. He, X. Sun, L. Li, X. Zhang, and H. Gan, “Activation
“PICK1: a perinuclear binding protein and substrate for pro- of sirtuin1 protects against ischemia/reperfusion-induced
tein kinase C isolated by the yeast two-hybrid system,” The acute kidney injury,” Biomedicine & pharmacotherapy = Bio-
Journal of Cell Biology, vol. 128, no. 3, pp. 263–271, 1995. médecine & pharmacothérapie, vol. 125, p. 110021, 2020.
[21] J. Staudinger, J. Lu, and E. N. Olson, “Specific interaction of the [37] X. Tian, H. Gan, Y. Zeng, H. Zhao, R. Tang, and Y. Xia, “Inhi-
PDZ domain protein PICK1 with the COOH terminus of pro- bition of semaphorin-3a suppresses lipopolysaccharide-
tein kinase C-α,” The Journal of Biological Chemistry, vol. 272, induced acute kidney injury,” Journal of Molecular Medicine,
no. 51, pp. 32019–32024, 1997. vol. 96, no. 7, pp. 713–724, 2018.
[22] J. Xu and J. Xia, “Structure and function of PICK1,” Neuro-Sig- [38] S. Sifuentes-Franco, D. E. Padilla-Tejeda, S. Carrillo-Ibarra,
nals, vol. 15, no. 4, pp. 190–201, 2006. and A. G. Miranda-Díaz, “Oxidative stress, apoptosis, and
[23] Y. N. Wang, L. Zhou, Y. H. Li et al., “Protein interacting with mitochondrial function in diabetic nephropathy,” Interna-
C-kinase 1 deficiency impairs glutathione synthesis and tional Journal of Endocrinology, vol. 2018, Article ID
increases oxidative stress via reduction of surface excitatory 1875870, 13 pages, 2018.
amino acid carrier 1,” The Journal of neuroscience : the official [39] K. Sinha, J. Das, P. B. Pal, and P. C. Sil, “Oxidative stress: the
journal of the Society for Neuroscience, vol. 35, no. 16, mitochondria-dependent and mitochondria-independent
pp. 6429–6443, 2015. pathways of apoptosis,” Archives of Toxicology, vol. 87, no. 7,
[24] M. Qian, Y. Lou, Y. Wang et al., “PICK1 deficiency exacerbates pp. 1157–1180, 2013.
sepsis-associated acute lung injury and impairs glutathione [40] Y. H. Li, N. Zhang, Y. N. Wang, Y. Shen, and Y. Wang, “Mul-
synthesis via reduction of xCT,” Free Radical Biology & Medi- tiple faces of protein interacting with C kinase 1 (PICK1):
cine, vol. 118, pp. 23–34, 2018. structure, function, and diseases,” Neurochemistry Interna-
[25] J. Xie, X. Wu, Q. Zhou et al., “PICK1 confers anti- tional, vol. 98, pp. 115–121, 2016.
inflammatory effects in acute liver injury via suppressing M1 [41] B. Lei, D. Wang, M. Zhang, Y. Deng, H. Jiang, and Y. Li, “miR-
macrophage polarization,” Biochimie, vol. 127, pp. 121–132, 615-3p promotes the epithelial-mesenchymal transition and
2016. metastasis of breast cancer by targeting PICK1/TGFBRI axis,”
[26] J. Y. Kim, S. J. Lee, Y. I. Maeng, J. Leem, and K. K. Park, “Pro- Journal of Experimental & Clinical Cancer Research, vol. 39,
tective effects of bee venom against endotoxemia-related acute no. 1, p. 71, 2020.
kidney injury in mice,” Biology, vol. 9, no. 7, p. 154, 2020. [42] Y. Mo, Y. Lou, A. Zhang et al., “PICK1 deficiency induces
[27] G. Wang, X. Zhang, X. Pan, and Y. Xiao, “FSC231 can alleviate autophagy dysfunction via lysosomal impairment and
paclitaxel-induced neuralgia by inhibiting PICK1 and affecting amplifies sepsis-induced acute lung injury,” Mediators of
related factors,” Neuroscience Letters, vol. 741, p. 135471, 2021. Inflammation, vol. 2018, Article ID 6757368, 11 pages, 2018.
[28] N. Robinson, R. Ganesan, C. Hegedűs, K. Kovács, T. A. Kufer, [43] Z. Luo, X. Xu, T. Sho et al., “ROS-induced autophagy regulates
and L. Virág, “Programmed necrotic cell death of macro- porcine trophectoderm cell apoptosis, proliferation, and differ-
phages: focus on pyroptosis, necroptosis, and parthanatos,” entiation,” American Journal of Physiology. Cell Physiology,
Redox Biology, vol. 26, p. 101239, 2019. vol. 316, no. 2, pp. C198–C209, 2019.
[29] A. Havasi and S. C. Borkan, “Apoptosis and acute kidney [44] Y. Luo, C. Wang, P. Yong et al., “Decreased expression of the
injury,” Kidney International, vol. 80, no. 1, pp. 29–40, 2011. long non-coding RNA SLC7A11-AS1 predicts poor prognosis
[30] S. M. Parikh, “Therapeutic targeting of the mitochondrial dys- and promotes tumor growth in gastric cancer,” Oncotarget,
function in septic acute kidney injury,” Current Opinion in vol. 8, no. 68, pp. 112530–112549, 2017.
Critical Care, vol. 19, no. 6, pp. 554–559, 2013. [45] W. L. Wang, S. F. Yeh, E. Y. Huang et al., “Mitochondrial
[31] H. Hu, M. Tian, C. Ding, and S. Yu, “The C/EBP homologous anchoring of PKCalpha by PICK1 confers resistance to
protein (CHOP) transcription factor functions in endoplasmic etoposide-induced apoptosis,” Apoptosis, vol. 12, no. 10,
reticulum stress-induced apoptosis and microbial infection,” pp. 1857–1871, 2007.
Frontiers in Immunology, vol. 9, p. 3083, 2019.
[32] C. Chen, W. Yao, S. Wu et al., “Crosstalk between connexin32
and mitochondrial apoptotic signaling pathway plays a pivotal
role in renal ischemia reperfusion-induced acute kidney
injury,” Antioxidants & Redox Signaling, vol. 30, no. 12,
pp. 1521–1538, 2019.
[33] Y. Gu, F. Huang, Y. Wang et al., “Connexin32 plays a crucial
role in ROS-mediated endoplasmic reticulum stress apoptosis
signaling pathway in ischemia reperfusion-induced acute kid-
ney injury,” Journal of Translational Medicine, vol. 16, no. 1,
p. 117, 2018.
[34] J. Kan, H. Huang, Z. Jiang et al., “Arenobufagin promoted oxi-
dative stress-associated mitochondrial pathway apoptosis in
A549 non-small-cell lung cancer cell line,” Evidence-Based

You might also like